The Aurora B Kinase Activity Is Required for the Maintenance of the Differentiated State of Murine Myoblasts

Total Page:16

File Type:pdf, Size:1020Kb

The Aurora B Kinase Activity Is Required for the Maintenance of the Differentiated State of Murine Myoblasts Cell Death and Differentiation (2009) 16, 321–330 & 2009 Macmillan Publishers Limited All rights reserved 1350-9047/09 $32.00 www.nature.com/cdd The Aurora B kinase activity is required for the maintenance of the differentiated state of murine myoblasts G Amabile1,2, AM D’Alise1, M Iovino1, P Jones3, S Santaguida4, A Musacchio4, S Taylor5 and R Cortese*,1 Reversine is a synthetic molecule capable of inducing dedifferentiation of C2C12, a murine myoblast cell line, into multipotent progenitor cells, which can be redirected to differentiate in nonmuscle cell types under appropriate conditions. Reversine is also a potent inhibitor of Aurora B, a protein kinase required for mitotic chromosome segregation, spindle checkpoint function, cytokinesis and histone H3 phosphorylation, raising the possibility that the dedifferentiation capability of reversine is mediated through the inhibition of Aurora B. Indeed, here we show that several other well-characterized Aurora B inhibitors are capable of dedifferentiating C2C12 myoblasts. Significantly, expressing drug-resistant Aurora B mutants, which are insensitive to reversine block the dedifferentiation process, indicating that Aurora B kinase activity is required to maintain the differentiated state. We show that the inhibition of the spindle checkpoint or cytokinesis per se is not sufficient for dedifferentiation. Rather, our data support a model whereby changes in histone H3 phosphorylation result in chromatin remodeling, which in turn restores the multipotent state. Cell Death and Differentiation (2009) 16, 321–330; doi:10.1038/cdd.2008.156; published online 31 October 2008 Lineage-restricted cells can be reprogramed to a state Evidence is emerging that the role of Aurora B is not of pluripotency by several different manipulations, including restricted to mitosis and cell division. A key substrate of somatic cell nuclear transfer1 embryonic stem cell (ES) Aurora B is serine 10 (ser10) of histone H3, and although this fusion 2 or even by cotransfection of four specific transcription phosphorylation event was originally thought to be involved in factors. 3–5 In particular, C2C12 myoblasts can be repro- chromosome condensation, it now appears that it dissociates gramed to dedifferentiate into multipotent progenitor cells HP1 proteins from methylated histone H3 (K9me3) at the that can be redirected to other cell types under appropriate onset of mitosis.13,14 Indeed, histone H3 (ser10) is an stimuli, including the ectopic expression of Msx1 gene,6 important element in the combinatorial histone code that is or by exposure to chemical compounds such as associated with both active and inactive chromatin.15 At the reversine.7 onset of mitosis, phospho-Ser10-H3 is responsible for the Reversine, a purine derivative, was identified by virtue of its ejection of HP1 proteins from their binding site on chromatin, ability to increase the plasticity of C2C12 myoblasts. This probably through a steric hindrance.13,14 Furthermore, it was compound interferes with the normal differentiation pathway demonstrated that the modifications of histone H3 and causing C2C12 cells to dedifferentiate to a pluripotent state. consequently the redistribution of HP1s are linked to More recently, it was demonstrated that reversine can chromatin reorganization and to cellular dedifferentation.16 differentiate human fibroblasts into skeletal muscle cells in Consistent with a role for Aurora B in chromatin structure, it vitro and in vivo,8 suggesting that reversine is capable of also was recently shown that Aurora B is required to remodel reprograming primary somatic cells to a state of increased chromatin during postmitotic cell differentiation of mesen- plasticity. We recently discovered that reversine is also a chymal stem cells and in the transition of B cell to plasma cells.17 potent Aurora kinase inhibitor.9 The Auroras are serine/ The ability of reversine to both dedifferentiate C2C12 cells threonine kinases required for multiple aspects of mitosis in and to inhibit Aurora B raised the possibility that its eukaryotic cells. Aurora A promotes centrosome maturation dedifferentiation capability is mediated through Aurora B. and spindle assembly,10 whereas Aurora B is required for Here, we present the evidence that in addition to reversine, histone H3 phosphorylation, chromosome segregation,11 the several other Aurora B inhibitors can dedifferentiate C2C12 spindle assembly checkpoint and cytokinesis.12 myoblasts. By using drug-resistant Aurora B mutants, we 1CEINGE Biotecnologie Avanzate, Naples, Italy; 2Department of Cellular Biotechnology and Hematology, University of Rome La Sapienza, Rome, Italy; 3IRBM Merck Research Laboratory, Pomezia, Italy; 4Department of Experimental Oncology, European Institute of Oncology, Milan, Italy and 5Faculty of Life Sciences, University of Manchester, Manchester, UK *Corresponding author: R Cortese, Medical Biotechnology, CEINGE Biotecnologie Avanzate, Via Comunale Margherita 482, Naples 80131, Italy. Tel: þ 39 335 81 04 832; Fax: þ 39 081 746 36 50; E-mail: [email protected] Keywords: dedifferentiation; reversine; Aurora B; myoblasts Abbreviations: ES, embryonic stem cells; ap2, adipocyte fatty acid-binding protein; LPL, lipoprotein lipase ; ALP, alkaline phosphatase; NMMII, nonmuscle myosin II heavy chain ; MEK, mitogen-activated extracellular signal-regulated kinase; ADM, adipogenic-differentiating medium; ODM, osteogenic-differentiating medium; MRFs, muscle regulatory factors; Id(s), inhibitor of differentiation(s); Myf5, myogenic factor 5; Lys, lysine; Ser, serine; p, phospho; me, methyl Received 18.6.08; revised 19.9.08; accepted 19.9.08; Edited by G Cossu; published online 31.10.08 Aurora B inhibition induces dedifferentiation G Amabile et al 322 provide compelling evidence that the effects of reversine are should have different spectra of off-target effects.19 We mediated through Aurora B as opposed to an off-target effect. chose to use hesperadin, which is relatively selective for Furthermore, we demonstrate that reversine remodels the Aurora B,20 VX-680, which is a dual Aurora A/B inhibitor21 chromatin associated with several genes that are induced or and MLN-8054, which is relatively selective for Aurora A.22 repressed during the phenomenon of dedifferentiation. Significantly, hesperadin, VX-680 and MLN-8054 induced Altogether, our data suggest a novel role for the Aurora B dedifferentiation of C2C12 myoblasts (Figure 1). C2C12 cells kinase in gene regulation and in the maintenance the were treated with compounds for 72 h as indicated; after this, differentiated state. the drug-containing media were removed and the cells were cultured in drug-free medium suitable for the development of adipocytes (adipogenic-differentiating medium; ADM) and Results and Discussion osteoblasts (osteogenic-differentiating medium; ODM), respectively. After 7 days, cells were stained with Oil Red Aurora kinase inhibitors reprogram C2C12 myo- O reagent to evaluate the presence of adipocytes or for blasts. Reversine inhibits several proteins including MEK1, alkaline phosphatase to evaluate the presence of osteoblasts nonmuscle myosin II (NMMII), Aurora A and B.9 On the basis (Figure 1a). In addition, to have a quantitative analysis of the of several indirect lines of evidence, it was suggested that dedifferentiation phenomenon, we used a fluorescent reversine induces dedifferentiation by the simultaneous substrate system to evaluate the amount of lipid droplets in inhibition of MEK1 and of NMMII.18 However, as reversine the case of adipocytes and the alkaline phosphatase activity is a potent Aurora inhibitor, we wanted to determine whether in the case of osteoblasts (Figure 1b, see Materials and the inhibition of Aurora A and/or B may be involved in the Methods also). dedifferentiation process. To do this, we asked whether a Furthermore, we decided to investigate also the expression panel of other Aurora kinase inhibitors could dedifferentiate of specific markers of both osteoblast and adipocyte C2C12 cells, reasoning that structurally diverse inhibitors differentiation. In particular, after the treatment with 500 nM ControlReversine VX-680 Hesperadin Oil Red staining ALP staining 72 hr treatment (500 nm) ALP activity Adipo Red assay 4500 4500 4000 10000 4000 12000 3500 3500 10000 3000 8000 3000 2500 8000 2500 6000 RFU 2000 2000 6000 1500 RFU 4000 1500 4000 1000 1000 2000 2000 500 500 0 0 0 0 Control VX-680 Control VX-680 Control VX-680 Control VX-680 Reversine Reversine Reversine Reversine Hesperadin MLN 8054 Hesperadin MLN 8054 Hesperadin MLN 8054 Hesperadin MLN 8054 72 hr treatment (500 nm) 72 hr treatment (50 nm) 72 hr treatment (500 nm) 72 hr treatment (50 nm) Figure 1 Aurora kinases inhibitors induce C2C12 dedifferentiation. (a) C2C12 cells were treated for 72 h with 500 nM of reversine, hesperadin and VX-680. Alkaline phoshatase and Oil Red O stainings were performed after 7 days of exposure to osteogenic-differentiating (ODM) and to adipogenic-differentiating media (ADM), respectively. The arrows indicate cells positive for Alkaline phosphatase (upper panel) and for lipid droplets content (lower panel) (b) C2C12 cells were treated for 72 h with 500 or 50 nM of reversine, hesperadin, VX-680 and MLN-8054. Then, cells were cultured in ODM and ADM media as indicated. ALP activity of C2C12 cells after the treatment was assayed using a fluorescent substrate system and the results were expressed in relative fluorescence units (RFUs). Quantitative analysis of adipocyte differentation was assayed with
Recommended publications
  • Aurora Kinase a in Gastrointestinal Cancers: Time to Target Ahmed Katsha1, Abbes Belkhiri1, Laura Goff3 and Wael El-Rifai1,2,4*
    Katsha et al. Molecular Cancer (2015) 14:106 DOI 10.1186/s12943-015-0375-4 REVIEW Open Access Aurora kinase A in gastrointestinal cancers: time to target Ahmed Katsha1, Abbes Belkhiri1, Laura Goff3 and Wael El-Rifai1,2,4* Abstract Gastrointestinal (GI) cancers are a major cause of cancer-related deaths. During the last two decades, several studies have shown amplification and overexpression of Aurora kinase A (AURKA) in several GI malignancies. These studies demonstrated that AURKA not only plays a role in regulating cell cycle and mitosis, but also regulates a number of key oncogenic signaling pathways. Although AURKA inhibitors have moved to phase III clinical trials in lymphomas, there has been slower progress in GI cancers and solid tumors. Ongoing clinical trials testing AURKA inhibitors as a single agent or in combination with conventional chemotherapies are expected to provide important clinical information for targeting AURKA in GI cancers. It is, therefore, imperative to consider investigations of molecular determinants of response and resistance to this class of inhibitors. This will improve evaluation of the efficacy of these drugs and establish biomarker based strategies for enrollment into clinical trials, which hold the future direction for personalized cancer therapy. In this review, we will discuss the available data on AURKA in GI cancers. We will also summarize the major AURKA inhibitors that have been developed and tested in pre-clinical and clinical settings. Keywords: Aurora kinases, Therapy, AURKA inhibitors, MNL8237, Alisertib, Gastrointestinal, Cancer, Signaling pathways Introduction stage [9-11]. Furthermore, AURKA is critical for Mitotic kinases are the main proteins that coordinate ac- bipolar-spindle assembly where it interacts with Ran- curate mitotic processing [1].
    [Show full text]
  • RASSF1A Interacts with and Activates the Mitotic Kinase Aurora-A
    Oncogene (2008) 27, 6175–6186 & 2008 Macmillan Publishers Limited All rights reserved 0950-9232/08 $32.00 www.nature.com/onc ORIGINAL ARTICLE RASSF1A interacts with and activates the mitotic kinase Aurora-A L Liu1, C Guo1, R Dammann2, S Tommasi1 and GP Pfeifer1 1Division of Biology, Beckman Research Institute, City of Hope Cancer Center, Duarte, CA, USA and 2Institute of Genetics, University of Giessen, Giessen, Germany The RAS association domain family 1A (RASSF1A) gene tumorigenesis and carcinogen-induced tumorigenesis is located at chromosome 3p21.3 within a specific area of (Tommasi et al., 2005; van der Weyden et al., 2005), common heterozygous and homozygous deletions. RASS- supporting the notion that RASSF1A is a bona fide F1A frequently undergoes promoter methylation-asso- tumor suppressor. However, it is not fully understood ciated inactivation in human cancers. Rassf1aÀ/À mice how RASSF1A is involved in tumor suppression. are prone to both spontaneous and carcinogen-induced The biochemical function of the RASSF1A protein is tumorigenesis, supporting the notion that RASSF1A is a largely unknown. The homology of RASSF1A with the tumor suppressor. However, it is not fully understood how mammalian Ras effector novel Ras effector (NORE)1 RASSF1A is involved in tumor suppression pathways. suggests that the RASSF1A gene product may function Here we show that overexpression of RASSF1A inhibits in signal transduction pathways involving Ras-like centrosome separation. RASSF1A interacts with Aurora-A, proteins. However, recent data indicate that RASSF1A a mitotic kinase. Surprisingly, knockdown of RASS- itself binds to RAS only weakly and that binding to F1A by siRNA led to reduced activation of Aurora-A, RAS may require heterodimerization of RASSF1A and whereas overexpression of RASSF1A resulted in in- NORE1 (Ortiz-Vega et al., 2002).
    [Show full text]
  • Structure-Based Discovery and Bioactivity Evaluation of Novel
    molecules Article Structure-Based Discovery and Bioactivity Evaluation of Novel Aurora-A Kinase Inhibitors as Anticancer Agents via Docking-Based Comparative Intermolecular Contacts Analysis (dbCICA) Majd S. Hijjawi 1 , Reem Fawaz Abutayeh 2 and Mutasem O. Taha 3,* 1 Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman 11942, Jordan; [email protected] 2 Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Applied Science Private University, Amman 11931, Jordan; [email protected] 3 Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman 11942, Jordan * Correspondence: [email protected]; Tel.: +962-6-535-5000 Academic Editors: Helen Osborn, Mohammad Najlah, Jean Jacques Vanden Eynde, Annie Mayence and Tien L. Huang Received: 15 October 2020; Accepted: 11 December 2020; Published: 18 December 2020 Abstract: Aurora-A kinase plays a central role in mitosis, where aberrant activation contributes to cancer by promoting cell cycle progression, genomic instability, epithelial-mesenchymal transition, and cancer stemness. Aurora-A kinase inhibitors have shown encouraging results in clinical trials but have not gained Food and Drug Administration (FDA) approval. An innovative computational workflow named Docking-based Comparative Intermolecular Contacts Analysis (dbCICA) was applied—aiming to identify novel Aurora-A kinase inhibitors—using seventy-nine reported Aurora-A kinase inhibitors to specify the best possible docking settings needed to fit into the active-site binding pocket of Aurora-A kinase crystal structure, in a process that only potent ligands contact critical binding-site spots, distinct from those occupied by less-active ligands. Optimal dbCICA models were transformed into two corresponding pharmacophores.
    [Show full text]
  • HIPK2 and Extrachromosomal Histone H2B Are Separately Recruited by Aurora-B for Cytokinesis
    Oncogene https://doi.org/10.1038/s41388-018-0191-6 ARTICLE HIPK2 and extrachromosomal histone H2B are separately recruited by Aurora-B for cytokinesis 1 1 2 3 2 Laura Monteonofrio ● Davide Valente ● Manuela Ferrara ● Serena Camerini ● Roberta Miscione ● 3 1,2 1 Marco Crescenzi ● Cinzia Rinaldo ● Silvia Soddu Received: 14 November 2017 / Revised: 24 January 2018 / Accepted: 5 February 2018 © The Author(s) 2018. This article is published with open access Abstract Cytokinesis, the final phase of cell division, is necessary to form two distinct daughter cells with correct distribution of genomic and cytoplasmic materials. Its failure provokes genetically unstable states, such as tetraploidization and polyploidization, which can contribute to tumorigenesis. Aurora-B kinase controls multiple cytokinetic events, from chromosome condensation to abscission when the midbody is severed. We have previously shown that HIPK2, a kinase involved in DNA damage response and development, localizes at the midbody and contributes to abscission by phosphorylating extrachromosomal histone H2B at Ser14. Of relevance, HIPK2-defective cells do not phosphorylate H2B 1234567890();,: and do not successfully complete cytokinesis leading to accumulation of binucleated cells, chromosomal instability, and increased tumorigenicity. However, how HIPK2 and H2B are recruited to the midbody during cytokinesis is still unknown. Here, we show that regardless of their direct (H2B) and indirect (HIPK2) binding of chromosomal DNA, both H2B and HIPK2 localize at the midbody independently of nucleic acids. Instead, by using mitotic kinase-specific inhibitors in a spatio-temporal regulated manner, we found that Aurora-B kinase activity is required to recruit both HIPK2 and H2B to the midbody.
    [Show full text]
  • Spatiotemporal Dynamics of Aurora B-PLK1-MCAK Signaling Axis
    www.nature.com/scientificreports OPEN Spatiotemporal dynamics of Aurora B-PLK1-MCAK signaling axis orchestrates kinetochore Received: 21 October 2014 Accepted: 13 May 2015 bi-orientation and faithful Published: 24 July 2015 chromosome segregation Hengyi Shao1, Yuejia Huang2,3, Liangyu Zhang1,3, Kai Yuan1, Youjun Chu1,3, Zhen Dou1,2, Changjiang Jin1, Minerva Garcia-Barrio3, Xing Liu1,2,3 & Xuebiao Yao1 Chromosome segregation in mitosis is orchestrated by the dynamic interactions between the kinetochore and spindle microtubules. The microtubule depolymerase mitotic centromere-associated kinesin (MCAK) is a key regulator for an accurate kinetochore-microtubule attachment. However, the regulatory mechanism underlying precise MCAK depolymerase activity control during mitosis remains elusive. Here, we describe a novel pathway involving an Aurora B-PLK1 axis for regulation of MCAK activity in mitosis. Aurora B phosphorylates PLK1 on Thr210 to activate its kinase activity at the kinetochores during mitosis. Aurora B-orchestrated PLK1 kinase activity was examined in real- time mitosis using a fluorescence resonance energy transfer-based reporter and quantitative analysis of native PLK1 substrate phosphorylation. Active PLK1, in turn, phosphorylates MCAK at Ser715 which promotes its microtubule depolymerase activity essential for faithful chromosome segregation. Importantly, inhibition of PLK1 kinase activity or expression of a non-phosphorylatable MCAK mutant prevents correct kinetochore-microtubule attachment, resulting in abnormal anaphase with chromosome bridges. We reason that the Aurora B-PLK1 signaling at the kinetochore orchestrates MCAK activity, which is essential for timely correction of aberrant kinetochore attachment to ensure accurate chromosome segregation during mitosis. During cell division, accurate chromosome segregation requires dynamic interactions between kineto- chores and spindle microtubules (MTs), which results in accurate chromosome bi-orientation1–4.
    [Show full text]
  • Mitotic Arrest Deficient 2 Expression Induces Chemosensitization to a DNA-Damaging Agent, Cisplatin, in Nasopharyngeal Carcinoma Cells
    Research Article Mitotic Arrest Deficient 2 Expression Induces Chemosensitization to a DNA-Damaging Agent, Cisplatin, in Nasopharyngeal Carcinoma Cells Hiu Wing Cheung,1 Dong-Yan Jin,2 Ming-tat Ling,1 Yong Chuan Wong,1 Qi Wang,1 Sai Wah Tsao,1 and Xianghong Wang1 Departments of 1Anatomy and 2Biochemistry, Faculty of Medicine, University of Hong Kong, Hong Kong, China Abstract mitotic checkpoint control, may be associated with tumorigenesis Recently, mitotic arrest deficient 2 (MAD2)–mediated spindle as well as cancer progression. Several regulators of the mitotic checkpoint have been identified checkpoint is shown to induce mitotic arrest in response to DNA damage, indicating overlapping roles of the spindle and most of them are localized to the kinetochore, which is checkpoint and DNA damage checkpoint. In this study, we connected to both the chromosome and the spindle (1). One of investigated if MAD2 played a part in cellular sensitivity to them, mitotic arrest deficient 2 (MAD2), is thought to be a key DNA-damaging agents, especially cisplatin, and whether it was component for a functional mitotic checkpoint because it is regulated through mitotic checkpoint. Using nine nasopha- required for generating the ‘‘wait’’ signal in response to microtubule ryngeal carcinoma (NPC) cell lines, we found that decreased disruption (1). Deletion or down-regulation of MAD2 leads to MAD2 expression was correlated with cellular resistance to mitotic checkpoint inactivation and chromosomal instability (4–6). cisplatin compared with the cell lines with high levels of Down-regulation of MAD2 has also been reported in human MAD2. Exogenous MAD2 expression in NPC cells also cancers such as lung (7), breast (8), nasopharyngeal (9), and ovarian conferred sensitivity to DNA-damaging agents especially carcinomas (10).
    [Show full text]
  • 1 Spindle Assembly Checkpoint Is Sufficient for Complete Cdc20
    Spindle assembly checkpoint is sufficient for complete Cdc20 sequestering in mitotic control Bashar Ibrahim Bio System Analysis Group, Friedrich-Schiller-University Jena, and Jena Centre for Bioinformatics (JCB), 07743 Jena, Germany Email: [email protected] Abstract The spindle checkpoint assembly (SAC) ensures genome fidelity by temporarily delaying anaphase onset, until all chromosomes are properly attached to the mitotic spindle. The SAC delays mitotic progression by preventing activation of the ubiquitin ligase anaphase-promoting complex (APC/C) or cyclosome; whose activation by Cdc20 is required for sister-chromatid separation marking the transition into anaphase. The mitotic checkpoint complex (MCC), which contains Cdc20 as a subunit, binds stably to the APC/C. Compelling evidence by Izawa and Pines (Nature 2014; 10.1038/nature13911) indicates that the MCC can inhibit a second Cdc20 that has already bound and activated the APC/C. Whether or not MCC per se is sufficient to fully sequester Cdc20 and inhibit APC/C remains unclear. Here, a dynamic model for SAC regulation in which the MCC binds a second Cdc20 was constructed. This model is compared to the MCC, and the MCC-and-BubR1 (dual inhibition of APC) core model variants and subsequently validated with experimental data from the literature. By using ordinary nonlinear differential equations and spatial simulations, it is shown that the SAC works sufficiently to fully sequester Cdc20 and completely inhibit APC/C activity. This study highlights the principle that a systems biology approach is vital for molecular biology and could also be used for creating hypotheses to design future experiments. Keywords: Mathematical biology, Spindle assembly checkpoint; anaphase promoting complex, MCC, Cdc20, systems biology 1 Introduction Faithful DNA segregation, prior to cell division at mitosis, is vital for maintaining genomic integrity.
    [Show full text]
  • Kinetochores, Microtubules, and Spindle Assembly Checkpoint
    Review Joined at the hip: kinetochores, microtubules, and spindle assembly checkpoint signaling 1 1,2,3 Carlos Sacristan and Geert J.P.L. Kops 1 Molecular Cancer Research, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands 2 Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands 3 Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands Error-free chromosome segregation relies on stable and cell division. The messenger is the SAC (also known as connections between kinetochores and spindle microtu- the mitotic checkpoint) (Figure 1). bules. The spindle assembly checkpoint (SAC) monitors The transition to anaphase is triggered by the E3 ubiqui- such connections and relays their absence to the cell tin ligase APC/C, which tags inhibitors of mitotic exit cycle machinery to delay cell division. The molecular (CYCLIN B) and of sister chromatid disjunction (SECURIN) network at kinetochores that is responsible for microtu- for proteasomal degradation [2]. The SAC has a one-track bule binding is integrated with the core components mind, inhibiting APC/C as long as incorrectly attached of the SAC signaling system. Molecular-mechanistic chromosomes persist. It goes about this in the most straight- understanding of how the SAC is coupled to the kineto- forward way possible: it assembles a direct and diffusible chore–microtubule interface has advanced significantly inhibitor of APC/C at kinetochores that are not connected in recent years. The latest insights not only provide a to spindle microtubules. This inhibitor is named the striking view of the dynamics and regulation of SAC mitotic checkpoint complex (MCC) (Figure 1).
    [Show full text]
  • MAD2 Expression in Oral Squamous Cell Carcinoma and Its Relationship to Tumor Grade and Proliferation
    ANTICANCER RESEARCH 34: 7021-7028 (2014) MAD2 Expression in Oral Squamous Cell Carcinoma and its Relationship to Tumor Grade and Proliferation CLARA RIZZARDI1, LUCIO TORELLI2, MANUELA SCHNEIDER3, FABIOLA GIUDICI4, LORENZO ZANDONA’1, MATTEO BIASOTTO5, ROBERTO DI LENARDA5 and MAURO MELATO6 1Unit of Pathology, Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy; 2Department of Mathematics and Earth Science, University of Trieste, Trieste, Italy; 3Unit of Pathology, ASS n.2 “Isontina”, Gorizia, Italy; 4Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy; 5Unit of Odontology and Stomatology, Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy; 6Scientific Research Institute and Hospital for Pediatrics “Burlo Garofolo”, Trieste, Italy Abstract. Background: Defects in the cell-cycle surveillance might contribute to the chromosomal instability observed in mechanism, called the spindle checkpoint, might contribute human cancers. Molecular analysis of the genes involved in to the chromosomal instability observed in human cancers, the spindle checkpoint has revealed relatively few genetic including oral squamous cell carcinoma. MAD2 and BUBR1 alterations, suggesting that the spindle checkpoint are key components of the spindle checkpoint, whose role in impairment frequently found in many human cancers might oral carcinogenesis and clinical relevance still need to be result from mutations in as yet unidentified checkpoint genes elucidated. Materials and Methods: We analyzed the or altered expression of known checkpoint genes. A better expression of MAD2 in 49 cases of oral squamous cell understanding of this mechanism might provide valuable carcinoma by immunohistochemistry and compared the insights into CIN and facilitate the design of novel findings with clinicopathological parameters, proliferative therapeutic approaches to treat cancer.
    [Show full text]
  • Bub1 Positions Mad1 Close to KNL1 MELT Repeats to Promote Checkpoint Signalling
    ARTICLE Received 14 Dec 2016 | Accepted 3 May 2017 | Published 12 June 2017 DOI: 10.1038/ncomms15822 OPEN Bub1 positions Mad1 close to KNL1 MELT repeats to promote checkpoint signalling Gang Zhang1, Thomas Kruse1, Blanca Lo´pez-Me´ndez1, Kathrine Beck Sylvestersen1, Dimitriya H. Garvanska1, Simone Schopper1, Michael Lund Nielsen1 & Jakob Nilsson1 Proper segregation of chromosomes depends on a functional spindle assembly checkpoint (SAC) and requires kinetochore localization of the Bub1 and Mad1/Mad2 checkpoint proteins. Several aspects of Mad1/Mad2 kinetochore recruitment in human cells are unclear and in particular the underlying direct interactions. Here we show that conserved domain 1 (CD1) in human Bub1 binds directly to Mad1 and a phosphorylation site exists in CD1 that stimulates Mad1 binding and SAC signalling. Importantly, fusion of minimal kinetochore-targeting Bub1 fragments to Mad1 bypasses the need for CD1, revealing that the main function of Bub1 is to position Mad1 close to KNL1 MELTrepeats. Furthermore, we identify residues in Mad1 that are critical for Mad1 functionality, but not Bub1 binding, arguing for a direct role of Mad1 in the checkpoint. This work dissects functionally relevant molecular interactions required for spindle assembly checkpoint signalling at kinetochores in human cells. 1 The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark. Correspondence and requests for materials should be addressed to G.Z.
    [Show full text]
  • The Closed Form of Mad2 Is Bound to Mad1 and Cdc20 at Unattached Kinetochores
    bioRxiv preprint doi: https://doi.org/10.1101/305763; this version posted April 21, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. The closed form of Mad2 is bound to Mad1 and Cdc20 at unattached kinetochores. Gang Zhang1,2,3 and Jakob Nilsson1 1 The Novo Nordisk Foundation Center for Protein Research, Faculty of health and medical sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark 2 Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266061, China 3 Qingdao Cancer Institute, Qingdao, Shandong 266061, China For correspondence: [email protected] or [email protected] Keywords: Mad2, Cdc20, Kinetochore, Spindle Assembly Checkpoint, Mad1 1 bioRxiv preprint doi: https://doi.org/10.1101/305763; this version posted April 21, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. ABSTRACT The spindle assembly checkpoint (SAC) ensures accurate chromosome segregation by delaying anaphase onset in response to unattached kinetochores. Anaphase is delayed by the generation of the mitotic checkpoint complex (MCC) composed of the checkpoint proteins Mad2 and BubR1/Bub3 bound to the protein Cdc20. Current models assume that MCC production is catalyzed at unattached kinetochores and that the Mad1/Mad2 complex is instrumental in the conversion of Mad2 from an open form (O-Mad2) to a closed form (C-Mad2) that can bind to Cdc20.
    [Show full text]
  • Discovery of BPR1K871, a Quinazoline Based
    www.impactjournals.com/oncotarget/ Oncotarget, 2016, Vol. 7, (No. 52), pp: 86239-86256 Research Paper Discovery of BPR1K871, a quinazoline based, multi-kinase inhibitor for the treatment of AML and solid tumors: Rational design, synthesis, in vitro and in vivo evaluation Yung Chang Hsu1,*, Mohane Selvaraj Coumar2,*, Wen-Chieh Wang1,*, Hui-Yi Shiao1,*, Yi-Yu Ke1, Wen-Hsing Lin1, Ching-Chuan Kuo1, Chun-Wei Chang1, Fu-Ming Kuo1, Pei-Yi Chen1, Sing-Yi Wang1, An-Siou Li1, Chun-Hwa Chen1, Po-Chu Kuo1, Ching- Ping Chen1, Ming-Hsine Wu1, Chen-Lung Huang1, Kuei-Jung Yen1, Yun-I Chang1, John T.-A. Hsu1, Chiung-Tong Chen1, Teng-Kuang Yeh1, Jen-Shin Song1, Chuan Shih1, Hsing-Pang Hsieh1,3 1Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC 2Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry, India 3Department of Chemistry, National Tsing Hua University, Hsinchu, Taiwan, ROC *These authors contributed equally to this work Correspondence to: Hsing-Pang Hsieh, email: [email protected] Keywords: acute myeloid leukemia, aurora kinase, FLT3, quinazoline, multi-kinase inhibitor Received: July 25, 2016 Accepted: November 07, 2016 Published: November 15, 2016 ABSTRACT The design and synthesis of a quinazoline-based, multi-kinase inhibitor for the treatment of acute myeloid leukemia (AML) and other malignancies is reported. Based on the previously reported furanopyrimidine 3, quinazoline core containing lead 4 was synthesized and found to impart dual FLT3/AURKA inhibition (IC50 = 127/5 nM), as well as improved physicochemical properties. A detailed structure-activity relationship study of the lead 4 allowed FLT3 and AURKA inhibition to be finely tuned, resulting in AURKA selective (5 and 7; 100-fold selective over FLT3), FLT3 selective (13; 30-fold selective over AURKA) and dual FLT3/AURKA selective (BPR1K871; IC50 = 19/22 nM) agents.
    [Show full text]